R e vie w

2 downloads 0 Views 1MB Size Report
diverse range of activities is a major challenge in research into melanoma initiation .... Acral lentiginous melanoma ... on the feet and, less commonly, the hands.
For reprint orders, please contact: [email protected]

Alejandro Conde-Perez1,2,3 & Lionel Larue*1,2,3 Institut Curie, Developmental Genetics of Melanocytes, Bat. 110, 91405, Orsay, France CNRS, UMR3347 Bat. 110, 91405, Orsay Cedex, France 3 INSERM, U1021 Bat. 110, 91405, Orsay Cedex, France *Author for correspondence: Tel.: +33 1 69 86 71 07 n Fax: +33 1 69 86 71 09 n [email protected] 1 2

The PI3K–PTEN–AKT signaling pathway is involved in various cellular activities, including proliferation, migration, cell growth, cell survival and differentiation during adult homeostasis as well as in tumorigenesis. It has been suggested that the constitutive activation of PI3K/AKT signaling with concurrent loss of function of the tumor suppressor molecule PTEN contributes to cancer formation. Members of the PI3K–PTEN–AKT pathway, including these proteins and mTOR, are altered in melanoma tumors and cell lines. A hallmark of activation of the pathway is the loss of function of PTEN. Indeed, loss of heterozygosity of PTEN has been observed in approximately 30% of human melanomas, implicating this signaling pathway in this cancer. PI3K signaling activation, via loss of PTEN function, can inhibit proapoptotic genes such as the FoxO family of transcription factors, while inducing cell growth- and cell survival-related elements such as p70S6K and AKT. Determining how the PI3K–PTEN–AKT signaling pathway, alone or in cooperation with other pathways, orchestrates the induction of target genes involved in a diverse range of activities is a major challenge in research into melanoma initiation and progression. Moreover, the acquisition of basic knowledge will help patient management with appropriate therapies that are already, or will shortly be, on the market.

Malignant melanoma is the most highly aggressive type of skin cancer, and the incidence of its diagnosis in westernized countries is increasing. Identification of the molecular mechanisms of melanoma formation (melanomagenesis) is imperative, as this cancer has a high propensity to metastasize and in many cases is resistant to currently available treatment. There has been extensive research undertaken to dissect and describe the key genetic and epigenetic alterations that result in aberrant signaling and subsequent malignancy. Several regulatory pathways play important roles in melanomagenesis including, but not limited to, MAPK, WNT and PI3K (Figure 1) . Although there are undoubtedly cooperative effects between these pathways, this issue is beyond the scope of this review. Constitutive activation of PI3K, via gain-of-function mutations in the PIK3CA gene encoding the class I PI3K catalytic subunit p110a, are observed in a variety of malignancies, from glioblastomas to lung cancers [1,2] , but only infrequently in melanoma. However, mutation, epigenetic and deletion events involving the tumor suppressor PTEN may be present in as many as 40–50% of sporadic melanomas [3] . Loss of PTEN function is a major contributing factor to increased signaling downstream from PI3K/AKT. A number of PI3K–PTEN–AKT cascade components have been identified, but their functions and regulation in the melanocyte lineage are still poorly 10.2217/FON.12.106 © 2012 Future Medicine Ltd

Review

Future Oncology

PTEN and melanomagenesis

understood. This is partly because most of the functional studies addressing PTEN function have used carcinoma, prostate and breast cancer models. Dissection of the PI3K–PTEN–AKT axis in melanoma should provide further insight into the melanocyte-specific molecular and cellular events involved in the initiation and/or progression of melanocytes towards melanoma. Melanomagenesis

Melanomagenesis is a multistep process that develops through a series of stages: n Common acquired nevi and dysplastic nevi; Radial growth phase melanoma;

n

Vertical growth phase melanoma;

n

Metastatic melanoma [4,5] . In a schematic way, considering cellular mechanisms, melanomagenesis may be defined as the initiation and progression of melanoma, in which initiation includes proliferation and bypassing senescence, and progression includes invasion and metastasis [6] . Invasion results from a combination of mechanisms: n

Pseudoepithelial-to-mesenchymal transition (melanocytes are not epithelial cells);

n

Keywords n

Loss of cell–cell adhesion;

n

n

catenin n LOH n MAPK melanoblasts n PI3K

Loss of cell–matrix adhesion;

n

Matrix degradation;

part of

n

Future Oncol. (2012) 8(9), 1109–1120

ISSN 1479-6694

1109

Review

Conde-Perez & Larue

HGF

IGF

WNT

FGF *

FRZ

RTK * RAS *

* RAF

PI3K

MEK

AKT

* *

ERK

*

* NF-κB

DSH

GSK3β

NF-κB *

CREB

* PTEN ∆

M-MITF

β-cat ∆

LEF p16

Figure 1. Oversimplified signaling pathways associated with PTEN. Three main signaling pathways (PI3K, MAPK and WNT/b‑catenin) are connected with PTEN. Some downstream targets are induced in normal conditions. Associated proteins can be mutated. Mutations are depicted as point mutations (*) or deletions (D).

Chemo-attraction/repulsion;

n

Migration.

n

Metastasis formation requires intravasation, extravasation, implantation at sites and subsequent angiogenesis. Typically, benign tumors remain ‘stuck’ at or before radial growth phase, and are proliferative but noninvasive. However, the acquisition of further genetic aberrations or epigenetic modifications often leads to a bypass of the senescence barrier and a phenotypic switch from proliferative to invasive. This is accompanied, during the later stages of the disease, by colonization of distal organs, including lung, liver and brain. Metastatic melanoma ultimately becomes highly resistant to all current forms of therapy. General clinical information

The most basic mode of early detection/identification, within the first stage, utilizes the ABCD guideline system [7] : Asymmetry;

n

Border irregularity;

n

Color;

n

Diameter.

n

1110

Future Oncol. (2012) 8(9)

This approach is widely acknowledged to be insufficient and misleading, as irregularity and heterogeneity varies within samples and not all relevant growths are necessarily identified [8,9] . Accurate diagnosis often requires a skin biopsy, which still remains the quickest method of detection [10] . The Breslow index is a prognostic factor for melanoma based on measuring the depth of tumor [11] . Monitoring the Breslow thickness in patients through time can be informative: higher values of the index are positively correlated with poor prognosis and can be used as an indicator for surgical removal. Currently, clinical evaluations for loss of PTEN involve a combination of sequencing and immunohistochemical techniques, with the latter being more effective in endometrial carcinomas [12] . These techniques can be readily applied to melanoma samples. Current therapy for melanoma

Although major improvements have been made in decoding the mechanisms involved in melanoma­g enesis, translation into clinical applications has unfortunately not been so effective. Over the last decade, melanoma research has focused on identifying possible therapeutic targets downstream of the R AS future science group

PTEN & melanomagenesis

signaling cascade and recently PI3K–PTEN– AKT, and seeking potential inhibitors. Two molecules, vemurafenib (PLX4032) and dabrafenib (GSK2118436), have been identified as inhibitors targeting BRAF mutations, specifically the V600E mutation. BRAF inhibitors have shown great promise in the clinic: 80% of patients with tumors associated with a V600E mutation in BRAF respond to the treatment. Unfortunately, resistance emerges approximately 6–9 months after the start of treatment with these agents. This resistance may be due to compensation by the MAPK and/or PI3K pathways. It has been shown that PTEN loss confers BRAF inhibitor resistance in melanoma cells [13] . In addition, resistance may also arise via a switch in signaling from BRAF to the CRAF isoform and subsequent hyperactivation of MEK–ERK [14] . In accordance, tumor samples obtained from melanoma patients treated with vemurafenib carry an activating mutation in MEK1 (C121S), which was absent from pretreated tumors; this mutation results in increased kinase activity and strong resistance to the inhibitor [15] . Similarly, mutation of MEK1 (K59del) decreases sensitivity to dabrafenib treatment in A375 human melanoma cells [16] . Thus, resistance to currently available BRAF inhibitors appears to result from a signaling switch between RAF isoforms and MEK1 mutations. Another therapeutic agent currently in use is the anti-CTLA4 antibody ipilimumab, with 10–20% of patients responding positively to the therapy. CKIT activity inhibitors may also be beneficial [17,18] . Inhibitors targeting functional components of the PI3K–PTEN–AKT pathway are available. The best characterized is rapamycin, which inhibits the mTORC1 complex and, at high concentrations, mTORC2. The mTOR complex is an attractive target for therapeutic exploitation as it regulates cell growth and survival. Treatment with rapamycin in in vitro cellular models leads to increased apoptosis, and decreased metastatic potential [19] . Currently, efforts are being made to develop isoform-specific mTOR inhibitors, in order to increase effectiveness while decreasing off-target effects [20] . Recent evidence has suggested that combinatorial therapy using BRAF inhibitors with mTOR inhibitors may be beneficial to bypass initial resistance to the BRAF inhibitor [21,22] . Various in vitro studies indicate that combinatorial therapies may provide the basis for new treatment strategies in the near future; nevertheless, the development of appropriate and effective regimens of this type for patients is still far from trivial. future science group

Review

Different types of melanoma

Melanoma exists in different forms. Superficial spreading melanoma accounts for approximately 40–70% of all cases of melanoma [23–26] . The most common locations are the legs of women and the backs of men, and they occur most commonly between 30 and 50 years of age. Many are barely raised from the surrounding skin and vary in color. Such melanomas evolve over 1–5 years and can be readily caught at an early stage if they are detected and removed. Nodular melanoma is the second most common subtype, and is described in 15% of melanoma cases [27] . It is vertically invasive and generally has a higher propensity to metastasize. They present as symmetrical nodules with colors ranging from blue, to brown, pink and grey (for review, see [28]). Lentigo maligna melanoma typically develops on the chronically sun-exposed skin of the head and neck [29] . Precursor lesions are termed lentigo maligna and commonly appear as irregular brownish pigmented macular lesions that persist for years. The incidence increases with age and generally peaks at between 70 and 80 years of age [30] . Acral lentiginous melanoma (ALM) is the rarest type of melanoma (~1% of malignant melanomas for the Australian population) and occurs at the same frequency in people of different phototypes [23] . It has a worse prognosis than the other types of melanoma [31–33] . Dermatological signs include dark, irregular macules, papules or nodules on the feet and, less commonly, the hands. Subungual occurrence in the fingers is uncommon, and is reported in 1–13% of all cases of ALM [34] . Histologically, ALM is characterized by asymmetric, poorly circumscribed proliferation of continuous single melanocytes at the dermal–epidermal junction [35] . The four subtypes display different histopathological features, diagnostic criteria and overall survival rates. Different genetic alterations are associated with different subtypes of the disease. The modification of PTEN may affect the development of superficial spreading melanoma, nodular melanoma and lentigo maligna melanoma, but does not seem to influence ALM [36] . Comparative genome hybridization analysis showed that chromosomal amplifications in regions 5p15, 5p13, 12q13–22 and 16q21–22 are more frequently associated with ALM and may drive tumor formation, irrespective of the status of 10q23 where PTEN is located [36] . However, this does not exclude that PTEN expression is altered in ALM. www.futuremedicine.com

1111

Review

Conde-Perez & Larue

Melanomagenesis: initiation & progression

The transformation of melanocytes, which are located in the skin and produce melanin pigments, leads to malignant melanoma. Both epigenetic and genetic alterations contribute to the etiology of melanoma. This includes genes corresponding to WNT/b‑catenin ( b‑catenin and APC), MAPK (BRAF and NRAS) and PI3K (PTEN and AKT3) signaling, cell adhesion (ITGB3, ITGAV and CDH1) and cell cycle control (CDKN2A or INK4A–ARF, MYC, RB1 and TP53) [37–39] . During the initial stage of melanomagenesis, abnormal melanocytic growth is observed in the form of nevi (moles). The cause of this abnormal proliferation is probably uncontrolled cell division and/or delayed senescence. Generally, once senescence occurs, these nevi do not progress towards malignancy. Mutated and hyperactive forms of NRAS and BR AF have been shown to be associated in melanocytes with proliferation and induction of senescence, respectively [40,41] . According to this, immortalization of the cells is required for melanoma to form. However, one must not forget that melanoma may still arise from a single melanocyte when both proliferation and immortalization events occur. In this case, it is possible that the molecular event associated with immortalization may occur first without any apparent lesion. In melanocytes, the MAPK (RAS/RAF/MEK/ERK) pathway is activated by growth factors, including SCF, HGF and FGF, resulting in ERK activation and thus proliferation [6,42] . BRAF is mutated in up to 70% of benign nevi and melanomas, leading to constitutive activation of the MAPK pathway [43] . Oncogenes such as BRAF (or NRAS) are themselves incapable of transforming primary cells, or can do so only poorly, as they cause oncogene-induced senescence [40,44,45] . Mutation and/or alteration of gene expression may lead to the immortalization of melanocytes. The P16/RB and P14/P53 pathways are classically involved in the bypass of senescence. In melanoma, the P16/RB pathway is particularly important in overcoming the senescence barrier to oncogenic stress, and P53 plays a larger role in melanomagenesis during melanoma progression (for review, see [46]). Deletion, mutation and silencing of P16 are the main molecular processes associated with the bypass of senescence in melanoma. In accordance, it has been shown that P16 can be silenced by repression by the b‑catenin–TCF4 complex or by methylation of its promoter [37] . Understanding how oncogenic 1112

Future Oncol. (2012) 8(9)

cells overcome this senescence barrier to hyperproliferate is one of the keys to improving our knowledge of melanoma initiation. PTEN

The gene encoding TEP1/MMAC1 was originally identified as a tumor suppressor, later on renamed PTEN due to its propensity to be mutated or lost in advanced cancers, resulting in a loss of function. It was formally proven as a tumor suppressor in vivo once its absence led to initiation of tumors. It shares significant similarity with the catalytic domain of protein phosphatases and the cytoskeletal proteins auxillin and tensin; consequently, it was classified as a dual protein and lipid phosphatase [47,48] . However, unlike other protein phosphatases, PTEN preferentially dephosphorylates phosphatidylinositol 3,4,5-trisphosphate (PIP3) at the 3´ position [49] , thereby negatively regulating the AKT signaling pathway. PTEN structure

The PTEN protein has 403 amino acids and two key functional domains: a phosphatase domain and a C2-spanning region regulating membrane stability [50] . It has an N-terminal phosphatidylinositol 3,4-bisphosphate (PIP2) binding region and a C-terminal PDZ domain regulating protein–protein interactions [51,52] . The N-terminal domain, the first 185 amino acids, contains a protein tyrosine phosphatase signature motif and shows structural similarity to the dual-specificity phosphatase VHR (F igur e  2) . However, crystallographic ana­lysis reveals that the residues corresponding to the active site in PTEN generate a larger groove for substrate binding than is observed for VHR, such that the substrate specificity is different [50] . The active site contains a HCXXGXXR amino acid signature commonly found in protein tyrosine phosphatases and dual specificity phosphatases [50] . The C2 domain (amino acids 186–352) contains a CBR3 loop differing slightly from those in PLCd1, PKCb and cPLA2, with only one calcium-binding residue (Asp286). Nevertheless, the loop has an overall net positive charge, resembling the Ca 2+ -dependent binding sites in the C2 domains of PLCd and PKCd; these structures may be involved in membrane binding [50] . The PIP2-binding region also contributes to the protein associating with the membrane [51] . Residues 401–403 in the PDZ-binding domain have been shown to mediate binding to MAG family kinases [52] . future science group

PTEN & melanomagenesis

1

15

PBM

185

Phosphatase domain

351

C2 domain

400

Reg domain

Review

403 AA

PDZ

Figure 2. PTEN structure. PTEN is composed of 403 AAs. It possesses five main domains or motifs. The plasma membrane-binding motif allows the binding of PTEN to PIP2. The phosphatase domain allows the dephosphorylation of PIP3 and, with lower efficiency, of proteins such as FAK. PTEN mutations found by deep sequencing (THR167ALA, CYS136TYR and TYR155CYS) map to this domain. The C2 domain allows the stabilization of PTEN to the membrane. The Reg domain, or regulatory domain, contains phosphorylable residues (Ser362, Thr366, Ser370 and Ser385) that are important for PTEN activity. PDZ allows the interaction with MAG family kinase. AA: Amino acid.

PTEN function

PTEN has been implicated in a plethora of cellular processes. A major role of PTEN is to dephosphorylate PIP3 resulting in an increase in the amount of PIP2 species and subsequently leading to a decrease in AKT signaling (reviewed in [53]). PIP2 is a major substrate of the PI3 family of kinases and its phosphorylation by PI3K, on the third hydroxyl group, results in production of the lipid second messenger, PIP3, which enables AKT binding to the membrane and subsequent phosphorylation of THR308 by PDK1 and SER473 by mTORC2 complex, leading to its activation [54,55] . This in turn leads to activation of the downstream signaling components of the PI3K–AKT cascade, ultimately modulating cellular processes including cell growth, cell size, cell cycle progression, cell survival, proliferation, growth and angiogenesis [56,57] . PTEN also has nuclear functions. In the majority of cutaneous melanomas PTEN may be found in the nucleus. Absence from the nucleus has been associated with poor prognosis [58] . The absence of PTEN from the nucleus has also been described in other cancers including colorectal cancer, pancreatic islet cell tumors and large B-cell lymphoma, leading to the view that the tumor suppressive activity of PTEN is not solely associated with its cytoplasmic functions [59–61] . Indeed, PTEN has been reported to be predominantly cytoplasmic in neoplastic tissue, although nuclear in normal tissue, suggesting the existence of a nuclear tumor-suppressing activity for this protein [62] . It is thought that nuclear PTEN is involved in cell cycle progression, chromatin stability and control of double-strand break DNA repair by interaction with the RAD51 promoter [63,64] . PTEN has been reported to interact directly with the promoter of P53, thereby increasing future science group

P53 transcript and protein levels [65] , although the role of the PTEN–P53 interaction is controversial as others have reported that PTEN inactivation leads to an increase of P53 production [66] . Nuclear PTEN is also involved in promoting APC–CDH1 complexing resulting in cellular senescence [67] . The upregulation of PTEN is also associated with an increase in energy expenditure and mitochondrial oxidative phosphorylation providing a protective effect against oncogenic transformation [68] . Regulation of PTEN

The gene for PTEN maps to the chromosomal region 10q23, and is often lost in various malignancies [49] . Loss of function of PTEN is often associated with an increase of PIP3 within the cells and subsequent activation of downstream AKT signaling. Indeed, re-expression in PTEN‑null cells leads to decreased cell proliferation and tumorigenicity [3] . PTEN activity is regulated by a multitude of processes, including post-translational acetylation by p300/CBP, which negatively affects its function [69] . Oxidative stress impairs PTEN catalytic activity by reducing cysteines 71 and 124, resulting in disulfide bonds [70–72] . NOTCH1 also regulates PTEN expression [73] . PTEN is mono- and poly-ubiquitinated by the E3 ubiquitin ligase, and NEDD4-1 has been reported to mediate its subcellular translocation and degradation, although this is still debated [74–76] . In several types of malignancy, PTEN expression is affected by miRNA 26a and 21, and the 106b‑25 miRNA cluster. Interestingly, the PTEN pseudogene PTENP1 was recently found to compete for miRNA targeting PTEN, thereby affecting the delicate balance of regulation [77] . Recently, PREX2 and MAGI2, known to interact with PTEN, were found to www.futuremedicine.com

1113

Review

Conde-Perez & Larue

be mutated [78] . The presence of these mutations may affect their interactions with PTEN. Functionally, it was previously shown that PREX2 negatively regulates the phospholipid activity of PTEN. However, it was not shown whether such mutation was effectively affecting PTEN activity when PREX2 was mutated. Lastly, PTEN activity is regulated by a series of phosphorylation events driven sequentially through CK2 and GSK3b [79–81] . These regulatory effects all have consequences for cellular processes that are associated with migration, proliferation, apoptosis, metabolism and the cell cycle. Commonly associated PTEN disorders in humans

PTEN mutations and deletions have been described in many human cancers including, but not limited to, prostate cancer, breast cancer and melanoma. Germline mutations have been found to be associated with Cowden’s, Lhermitte–Duclos and Bannayan–Zonana syndromes resulting in increased susceptibility to developing breast and nonmedullary thyroid carcinoma, dysplastic gliocytoma and endometrial carcinoma [82,83] . Over 80% of patients diagnosed with Cowden’s syndrome carry mutations in the PTEN gene [84] . The most common symptom associated with Cowden’s syndrome is the presence of multiple hamartomas in various organs [83] . Patients with Lhermitte–Duclos syndrome present obstructive hydrocephalus and mass effects, characterized by focal enlargements of the cerebellar folia [85] . Similarly, patients with Bannayan–Zonana syndrome exhibit cephalic abnormalities, mainly macrocephaly, as well as vascular malformations, subcutaneous and visceral lipoma, and intestinal hamartomas [86] . The most frequently mutated site (in ~30% of cases) in the PTEN gene is exon 5, which encodes the phosphatase domain [82] . Somatic mutations have been reported in a large number of cases of endometrial carcinoma and glioblastomas, and with varying degrees of frequency in prostate, breast, ovarian and skin cancers [82] . Involvement of PTEN in melanocyte transformation

The knowledge acquired regarding the development and the establishment of the melanocyte lineage is very informative about melanoma­ genesis, although it only provides a deforming mirror image [87] . At the molecular level, this cumulative knowledge is certainly true for any 1114

Future Oncol. (2012) 8(9)

gene/protein, including PTEN. For instance, cells move in the organism during both embryonic and cancer development. For oncologists, the movement of a ‘transformed’ cell in the organism is not spatially and temporally controlled by this cell itself, and is discordant; consequently, the term ‘invasion’ may be better adapted. For embryologists, the movement of a cell in the organism has two characteristics: it is spatially and temporally controlled and is in harmony with the other cells. As a result, the term ‘colonization’ is generally used. Despite these differences, both ‘colonization’ and ‘invasion’ result from combinations of different mechanisms: loss of cell–cell adhesion, loss of cell–matrix adhesion, matrix degradation, chemo-attraction/ repulsion and migration. The PI3K pathway is clearly involved in these different cellular mechanisms [88–91] . For instance, it has been shown that AKT regulates E-cadherin expression, therefore it is important for cell–cell adhesion [88] . Molecular oncologists often refer to invasion as observed in tests in vitro using various matrices, for example Matrigel™ (BD Biosciences, NJ, USA). Obviously this test, although very useful, only partly reflects the full invasion process in vivo, as it addresses mainly matrix degradation and chemo-attraction/repulsion in  vitro. During the establishment of the melanocyte lineage, embryologists refer to migration and not to colonization. This is because the loss of cell–cell adhesion, loss of cell–matrix adhesion and matrix degradation do not seem to be important at this stage: the tissues are very ‘loose’ until the end of organogenesis (~E14.5) and become tighter during fetal development. Consequently, migration remains the main cellular mechanism for melanoblast movements until that period. PTEN in the establishment of the melanocyte lineage

The exact role of PTEN in embryonic development has not been clearly elucidated, but experiments in mice showed that it is clearly important as homozygous loss leads to embryonic lethality at the time of gastrulation. The role of PTEN in the establishment of the melanocyte lineage is also not known, but the absence of this protein does not lead to major coat color phenotype in mice [92] , and PTEN is produced in murine melanoblasts [93] . Interestingly, after backcrossing the mice more than ten-times on C57BL/6 the hyperpigmentation in the tail, pinna, paws, skin and olfactory bulb in HM mice disappeared [Puig I et al., Unpublished Data] . At this point, the involved modifier gene(s) remain unknown. future science group

PTEN & melanomagenesis

PTEN is absent in a large proportion of melanomas

Various mutations in the PTEN gene have been identified in cutaneous melanoma: the frequency of their occurrence is reported to be approximately 10% for primary melanoma and 40% for melanoma cell lines [57] . Several groups have identified somatic mutations occurring at the PTEN locus in primary and metastatic melanoma with a higher frequency in metastatic samples [94,95] . Mutations can occur all throughout the PTEN coding region. The most common genetic alteration of PTEN in melanoma, associated with loss of function and melanoma development, is loss of heterozygosity [96] . Recently, three independent deep-sequencing studies were performed, two in melanoma cell lines and one on melanoma metastasis [78,97,98] . Besides the frequent chromosomal rearrangement, loss of heterozygosity of the region and the classical PTEN deletion, PTEN can present some point mutations (THR167ALA, CYS136TYR and TYR155CYS) in which phosphorylation of PTEN can be potentially affected. Role of PTEN in melanomagenesis

To elucidate the role of PTEN in melanoma initiation and progression, various in vitro and in vivo models have been established and characterized. Such models can also be used to screen drugs and new therapies. The constitutive inactivation of the Pten gene in mice leads to death at embryonic day 9.5 when homozygous. Heterozygous mice are viable and fertile but develop a large range of tumor types, except melanoma. The importance of PTEN in melanomagenesis was assessed using constitutively heterozygous mice and conditionally mutated mice for this gene. Early on, mice mutated for Pten, as heterozygous, and Ink4A/Arf, as homozygous, produced cutaneous melanoma [99] . These results provided the first evidence that PTEN was a cause of melanoma initiation. However, this mouse model was far from optimal for the study of melanoma; the penetrance was low (three out of 46), the latency period was long (~7 months) and, more importantly, many other types of tumor arose earlier (from 7 weeks of age) and more frequently (in ~75%). Mouse molecular genetic technology has allowed the generation of conditional mouse mutants for Pten using the CRE–LoxP system [6] . To generate melanocyte-specific transgenic expression, regulatory sequences of tyrosinase family genes, such as those from Tyr itself, Dct and Tyrp1 are used by many laboratories [100] . The Tyr promoter has been manipulated for future science group

Review

better expression in transgenic mice [101,102] . Subsequently, a combinatorial approach of TYR enhancer and Tyr promoter sequence was used to generate transgenic mouse lines enabling a melanocyte-specific expression of CRE recombinase (TYR::CRE, spatial control) [103,104] and tamoxifen-inducible expression of CRE recombinase (TYR::CREERt2, spatio-temporal control) [105,106] . The expression of CRE recombinase was also successful in the melanocyte lineage after insertion of CRE into the Dct locus, although expression is weaker than that from TYR::CRE constructs [107] . The inactivation of Pten in the melanocyte lineage, and some neural crest derivatives, using the DCT::CRE mice led to 50% lethality after birth with the remaining population having increased melanocyte counts in the skin and displaying susceptibility to carcinogen-induced melanomagenesis [108] . Even though DCT::CRE mice are not efficiently recombining floxed genes present in melanocytes, the specific inactivation of PTEN in the melanocyte lineage leads to a resistance to hair graying [108] . These results suggest that PTEN is important in the renewal of melanocytes. Several explanations can be given. In the absence of PTEN the number of melanocyte stem cells in each bulge is more important; melanocyte stem cells overcome natural senescence or transit amplifying cells divide more than normal for each cycle. It is widely accepted that NRAS activates both MAPK and PI3K pathways, but BRAF activates only the MAPK pathway. Conditional inactivation of PTEN and activation of BRAF in mice after birth has been used to evaluate synergy between the MAPK and PI3K signaling pathways. These mutant mice formed melanoma with short latency and 100% penetrance. Metastasis to distal organs, lung and lymph nodes was observed [109] . The conditional expression of a stabilized form of b‑catenin in a BRAFV600E, PTEN-null context led to the acceleration of melanoma initiation and increased metastasis [42,110] . It has also been shown that PTEN ceRNA cooperates with BRAF V600E to promote melanomagenesis, providing further evidence of the importance of PTEN regulation in melanoma formation [111] . At the molecular level, it is important to note that PTEN deficiency can cooperate with HRASV12G hyperactivation to promote murine melanomagenesis [112] . The relevance for human melanoma is questionable as HRAS mutations are found in only 1% of cases, and concomitant mutation of RAS (N, K or H) has not been found associated www.futuremedicine.com

1115

Review

Conde-Perez & Larue

with PTEN deficiency in melanoma biopsies. However, it has to be noted that NRAS and PTEN deficiency were reported in melanoma cells in culture [38] . Efforts are currently being made to generate mice that accurately mimic human melanoma formation to allow further characterization of the effects of PTEN function and its role in the disease. One potential approach could be the generation of mice that can be conditionally and temporally repressed for PTEN at different periods of melanoma initiation and progression using, for instance, the tetracyclin system. The strict comparison of these different mouse melanoma models is difficult because they present many differences including the genetic background. This last parameter was shown to be very important for melanoma initiation [113] . Unfortunately, there are no available data concerning progression in which invasion and metastasis are involved. Conclusion

The available evidence suggests that PTEN exerts its tumor-suppressive effects via several mechanisms. Whether it is through senescence bypass, downregulating survival kinases or by interacting with chromatin, it is clear that PTEN is an important tumor suppressor in melanoma. Premalignant lesions contain PTEN but often also carry a hyperproliferative mutation in the form of BRAF V600E . The exact mechanism of tumorigenic induction is not known but the evidence implicating PTEN is increasing. As a therapeutic target, PTEN seems an unlikely candidate due to its propensity to be lost in malignant melanoma. However, developing strategies to target downstream components, such as mTOR, with small-molecule inhibitors may be an option to compensate for the loss of PTEN

function; such inhibitors, used in combination with other small-molecule inhibitors, may confer a survival advantage for patients. Further studies are needed to fully appreciate the role of PTEN in melanoma­genesis and the importance of its numerous downstream effectors, which may or may not be post-translationally modified during the process. Work in this area should lead to an evaluation of the usefulness of some of these effectors both as biomarkers and therapeutic targets. Future perspective

The better knowledge of the structure and function of PTEN will allow discovery of its numerous interactors, and the functions of these various complexes. Such knowledge will lead to the better control of such interactions. Affecting these interactions will modulate the function of the specific complex. Another challenge will certainly be to understand the temporal importance of these different complexes. The temporal repression of PTEN at different periods of melanoma initiation and progression using, for instance, the tetra­cyclin system would certainly inform us on the role of this protein during melanomagenesis.u Financial & competing interests disclosure

A Conde-Perez was supported by a fellowship from Institut Curie. This work was supported by the Ligue Nationale Contre le Cancer (Equipe labellisée), Institut National sur le Cancer and Association de la Recherche sur le Cancer.��������������������������������������� The authors have no other relevant affili‑ ���� ations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.

Executive summary Structure & function „„PTEN is a phosphatase (lipid and protein) primarily found in the cytoplasm, although it can also be found in the nucleus. PTEN regulates its own phosphatase activity by appropriate folding of the protein, when it is properly post-translationally modified. Molecular biology „„In melanoma, PTEN is found to be mutated (most frequently on exon 5) in 17% of melanoma cases. PTEN is deleted in 13% of melanoma cases. Human genetic diseases „„PTEN may be altered in various syndromes including Cowden’s, Lhermitte–Duclos and Bannayan–Zonana, resulting in increased susceptibility to developing breast and nonmedullary thyroid carcinoma, dysplastic gliocytoma and endometrial carcinoma, but not melanoma. PTEN mouse models „„Constitutive and conditional PTEN mutations have revealed the importance of this protein during homeostasis and melanomagenesis. The lack of PTEN in melanocyte stem cells renders them more resistant to hair graying. The presence of BRAF (V600E) as well as HRAS (G12V) mutations along with the absence of PTEN in melanocytes is sufficient to initiate melanomagenesis.

1116

Future Oncol. (2012) 8(9)

future science group

PTEN & melanomagenesis

References Papers of special note have been highlighted as: n of interest nn of considerable interest

suppression of BIM expression. Cancer Res. 71(7), 2750–2760 (2011). 14. Heidorn SJ, Milagre C, Whittaker S et al.

Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 140(2), 209–221 (2010).

1.

Gymnopoulos M, Elsliger MA, Vogt PK. Rare cancer-specific mutations in PIK3CA show gain of function. Proc. Natl Acad. Sci. USA 104(13), 5569–5574 (2007).

2.

Samuels Y, Wang Z, Bardelli A et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 304(5670), 554 (2004).

3.

Stahl JM, Cheung M, Sharma A, Trivedi NR, Shanmugam S, Robertson GP. Loss of PTEN promotes tumor development in malignant melanoma. Cancer Res. 63(11), 2881–2890 (2003).

15. Wagle N, Emery C, Berger MF et al.

Elder DE, Guerry DT, Epstein MN et al. Invasive malignant melanomas lacking competence for metastasis. Am. J. Dermatopathol. 6(Suppl. 1), 55–61 (1984).

16. Greger JG, Eastman SD, Zhang V et al.

4.

5.

6.

7.

8.

9.

Herlyn M, Thurin J, Balaban G et al. Characteristics of cultured human melanocytes isolated from different stages of tumor progression. Cancer Res. 45(11 Pt 2), 5670–5676 (1985). Larue L, Beermann F. Cutaneous melanoma in genetically modified animals. Pigment Cell Res. 20(6), 485–497 (2007). Friedman RJ, Rigel DS, Kopf AW. Early detection of malignant melanoma: the role of physician examination and self-examination of the skin. CA Cancer J. Clin. 35(3), 130–151 (1985). Aldridge RB, Zanotto M, Ballerini L, Fisher RB, Rees JL. Novice identification of melanoma: not quite as straightforward as the ABCDs. Acta Derm. Venereol. 91(2), 125–130 (2011). Salerni G, Lovatto L, Carrera C, Puig S, Malvehy J. Melanomas detected in a follow-up program compared with melanomas referred to a melanoma unit. Arch. Dermatol. 147(5), 549–555 (2011).

n

Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J. Clin. Oncol. 29(22), 3085–3096 (2011). Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol. Cancer Ther. 11(4), 909–920 (2012). 17. Martí RM, Sorolla A, Yeramian A. New

therapeutic targets in melanoma. Actas Dermosifiliogr. 103(7), 579–590 (2012). 18. Robert C, Mateus C. Anti-CTLA-4

monoclonal antibody: a major step in the treatment of metastatic melanoma. Med. Sci. (Paris) 27(10), 850–858 (2011). 19. Yang Z, Lei Z, Li B et al. Rapamycin inhibits

lung metastasis of B16 melanoma cells through down-regulating alphav integrin expression and up-regulating apoptosis signaling. Cancer Sci. 101(2), 494–500 (2010). 20. Espona-Fiedler M, Soto-Cerrato V, Hosseini

A et al. Identification of dual mTORC1 and mTORC2 inhibitors in melanoma cells: prodigiosin vs. obatoclax. Biochem. Pharmacol. 83(4), 489–496 (2012). 21. Deng W, Gopal YN, Scott A, Chen G,

Woodman SE, Davies MA. Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition. Pigment Cell Melanoma Res. 2, 248–258 (2012).

10. Rigel DS, Russak J, Friedman R. The

evolution of melanoma diagnosis: 25 years beyond the ABCDs. CA Cancer J. Clin. 60(5), 301–316 (2010). 11. Shenenberger DW. Cutaneous malignant

22. Atefi M, Von Euw E, Attar N et al. Reversing

melanoma cross-resistance to BRAF and MEK inhibitors by co-targeting the AKT/ mTOR pathway. PLoS ONE 6(12), e28973 (2011)

melanoma: a primary care perspective. Am. Fam. Physician 85(2), 161–168 (2012). 12. Djordjevic B, Hennessy BT, Li J et al.

Clinical assessment of PTEN loss in endometrial carcinoma: immunohistochemistry outperforms gene sequencing. Mod. Pathol. 25, 699–708 (2012). 13. Paraiso KH, Xiang Y, Rebecca VW et al.

PTEN loss confers BRAF inhibitor resistance to melanoma cells through the

future science group

Shows that the action of a specific inhibitor of a mutated form of BRAF (BRAF V600E ) leads to a resistance mediated by the activation of proteins of the same signaling pathway.

n

Shows the importance of targeting several signaling pathways in order to bypass some resistance associated with one single signaling pathway.

23. Boniol M, Armstrong BK, Doré JF. Variation

in incidence and fatality of melanoma by season of diagnosis in New South Wales,

www.futuremedicine.com

Review

Australia. Cancer Epidemiol. Biomarkers Prev. 15(3), 524–526 (2006). 24. Mcpherson M, Elwood M, English DR,

Baade PD, Youl PH, Aitken JF. Presentation and detection of invasive melanoma in a high-risk population. J. Am. Acad. Dermatol. 54(5), 783–792 (2006). 25. Tournillac I, Picot MC, Dereure O, Guilhou

JJ, Guillot B. [Dubreuilh melanoma: and epidemiologic and prognostic study]. Ann. Dermatol. Venereol. 126(10), 676–680 (1999). 26. Saxe N, Hoffman M, Krige JE, Sayed R,

King HS, Hounsell K. Malignant melanoma in Cape Town, South Africa. Br. J. Dermatol. 138(6), 998–1002 (1998). 27. Chang AE, Karnell LH, Menck HR.

The National Cancer Data Base report on cutaneous and noncutaneous melanoma. A summary of 84,836 cases from the past decade. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer 83(8), 1664–1678 (1998). 28. Moloney FJ, Menzies SW. Key points in the

dermoscopic diagnosis of hypomelanotic melanoma and nodular melanoma. J. Dermatol. 38(1), 10–15 (2011). 29. Clark WH Jr, From L, Bernardino EA,

Mihm MC. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer Res. 29(3), 705–727 (1969). 30. Situm M, Bolanca Z, Buljan M. Lentigo

maligna melanoma – the review. Coll. Antropol. 34(Suppl. 2), 299–301 (2010). 31. Bradford PT, Goldstein AM, McMaster ML,

Tucker MA. Acral lentiginous melanoma: incidence and survival patterns in the United States, 1986–2005. Arch. Dermatol. 145(4), 427–434 (2009). 32. Swan MC, Hudson DA. Malignant

melanoma in South Africans of mixed ancestry: a retrospective analysis. Melanoma Res. 13(4), 415–419 (2003). 33. Bellows CF, Belafsky P, Fortgang IS, Beech

DJ. Melanoma in African–Americans: trends in biological behavior and clinical characteristics over two decades. J. Surg. Oncol. 78(1), 10–16 (2001).t 34. Miranda BH, Haughton DN, Fahmy FS.

Subungual melanoma: an important tip. J. Plast. Reconstr. Aesthet. Surg. doi:10.1016/j. bjps.2012.03.001 (2012) (Epub ahead of print). 35. Piliang MP. Acral lentiginous melanoma.

Clin. Lab. Med. 31(2), 281–288 (2011). 36. Bastian BC, Kashani-Sabet M, Hamm H

et al. Gene amplifications characterize acral melanoma and permit the detection of occult

1117

Review

Conde-Perez & Larue

tumor cells in the surrounding skin. Cancer Res. 60(7), 1968–1973 (2000). 37. Delmas V, Beermann F, Martinozzi S et al.

Beta-catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev. 21(22), 2923–2935 (2007). n

Shows that the cooperation of NRAS and b‑catenin induces melanoma formation from cells located in the bulge of the hair follicle containing normal melanocyte stem cells and transit amplifying cells. It also reveals that the PI3K pathway is induced.

38. Tsao H, Zhang X, Fowlkes K, Haluska FG.

Relative reciprocity of NRAS and PTEN/ MMAC1 alterations in cutaneous melanoma cell lines. Cancer Res. 60(7), 1800–1804 (2000). 39. Hayward NK. Genetics of melanoma

predisposition. Oncogene 22(20), 3053–3062 (2003). 40. Ackermann J, Frutschi M, Kaloulis K,

Mckee T, Trumpp A, Beermann F. Metastasizing melanoma formation caused by expression of activated N-RasQ61K on an INK4a-deficient background. Cancer Res. 65(10), 4005–4011 (2005). 41. Gray-Schopfer VC, Cheong SC, Chong H

et al. Cellular senescence in naevi and immortalisation in melanoma: a role for p16? Br. J. Cancer 95(4), 496–505 (2006). 42. Halaban R. Growth factors and melanomas.

Semin. Oncol. 23(6), 673–681 (1996). 43. Garnett MJ, Marais R. Guilty as charged:

B-RAF is a human oncogene. Cancer Cell 6(4), 313–319 (2004). 44. Michaloglou C, Vredeveld LC, Soengas MS

et al. BRAFE600 -associated senescence-like cell cycle arrest of human naevi. Nature 436(7051), 720–724 (2005). 45. Chin L, Tam A, Pomerantz J et al. Essential

role for oncogenic Ras in tumour maintenance. Nature 400(6743), 468–472 (1999). 46. Ha L, Merlino G, Sviderskaya EV.

Melanomagenesis: overcoming the barrier of melanocyte senescence. Cell Cycle 7(13), 1944–1948 (2008). 47. Li J, Yen C, Liaw D et al. PTEN, a putative

protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275(5308), 1943–1947 (1997). 48. Steck PA, Pershouse MA, Jasser SA et al.

Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q233 that is mutated in multiple advanced cancers. Nat. Genet. 15(4), 356–362 (1997).

1118

49. Simpson L, Parsons R. PTEN: life as a

61. Fridberg M, Servin A, Anagnostaki L et al.

tumor suppressor. Exp. Cell Res. 264(1), 29–41 (2001).

Protein expression and cellular localization in two prognostic subgroups of diffuse large B-cell lymphoma: higher expression of ZAP70 and PKC-beta II in the non-germinal center group and poor survival in patients deficient in nuclear PTEN. Leuk. Lymphoma 48(11), 2221–2232 (2007).

50. Lee JO, Yang H, Georgescu MM et al.

Crystal structure of the PTEN tumor suppressor: implications for its phosphoinositide phosphatase activity and membrane association. Cell 99(3), 323–334 (1999).

62. Planchon SM, Waite KA, Eng C. The

nuclear affairs of PTEN. J. Cell Sci. 121(Pt 3), 249–253 (2008).

51. Rahdar M, Inoue T, Meyer T, Zhang J,

Vazquez F, Devreotes PN. A phosphorylation-dependent intramolecular interaction regulates the membrane association and activity of the tumor suppressor PTEN. Proc. Natl Acad. Sci. USA 106(2), 480–485 (2009).

63. Chung JH, Ostrowski MC, Romigh T,

Minaguchi T, Waite KA, Eng C. The ERK1/2 pathway modulates nuclear PTEN-mediated cell cycle arrest by cyclin D1 transcriptional regulation. Hum. Mol. Genet. 15(17), 2553–2559 (2006).

52. Wu X, Hepner K, Castelino-Prabhu S et al.

Evidence for regulation of the PTEN tumor suppressor by a membrane-localized multi-PDZ domain containing scaffold protein MAGI-2. Proc. Natl Acad. Sci. USA 97(8), 4233–4238 (2000).

64. Shen WH, Balajee AS, Wang J et al.

Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 128(1), 157–170 (2007). n

53. Knobbe CB, Lapin V, Suzuki A, Mak TW.

The roles of PTEN in development, physiology and tumorigenesis in mouse models: a tissue-by-tissue survey. Oncogene 27(41), 5398–5415 (2008). 54. Sarbassov DD, Guertin DA, Ali SM,

Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor–mTOR complex. Science 307(5712), 1098–1101 (2005). 55. Hresko RC, Mueckler M. mTOR.RICTOR

65. Freeman DJ, Li AG, Wei G et al. PTEN

tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms. Cancer Cell 3(2), 117–130 (2003). 66. Chen Z, Trotman LC, Shaffer D et al.

Crucial role of p53-dependent cellular senescence in suppression of PTEN-deficient tumorigenesis. Nature 436(7051), 725–730 (2005).

is the Ser473 kinase for Akt/protein kinase B in 3T3-L1 adipocytes. J. Biol. Chem. 280(49), 40406–40416 (2005). 56. Salmena L, Carracedo A, Pandolfi PP. Tenets

of PTEN tumor suppression. Cell 133(3), 403–414 (2008).

67. Song MS, Carracedo A, Salmena L et al.

Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphataseindependent manner. Cell 144(2), 187–199 (2011).

57. Aguissa-Toure AH, Li G. Genetic alterations

of PTEN in human melanoma. Cell. Mol. Life Sci. 69(9), 1475–1491 (2012). 58. Whiteman DC, Zhou XP, Cummings MC,

Pavey S, Hayward NK, Eng C. Nuclear PTEN expression and clinicopathologic features in a population-based series of primary cutaneous melanoma. Int. J. Cancer. 99(1), 63–67 (2002). 59. Zhou XP, Loukola A, Salovaara R et al.

PTEN mutational spectra, expression levels, and subcellular localization in microsatellite stable and unstable colorectal cancers. Am. J. Pathol. 161(2), 439–447 (2002). 60. Perren A, Komminoth P, Saremaslani P et al.

Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared with normal islet cells. Am. J. Pathol. 157(4), 1097–1103 (2000).

Future Oncol. (2012) 8(9)

Demonstrates that PTEN interacts directly with chromatin-related protein CENAP, which is present in the nucleus and important in the regulation of chromosomal integrity.

nn

Shows that PTEN interacts with APC/C and promotes the formation of this complex with CDH1 (Fzr1 and not E-cadherin), which is a haploinsufficient tumor suppressor. This interaction leads to cell cycle division.

68. Garcia-Cao I, Song MS, Hobbs RM et al.

Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 149(1), 49–62 (2012). 69. Okumura K, Mendoza M, Bachoo RM,

Depinho RA, Cavenee WK, Furnari FB. PCAF modulates PTEN activity. J. Biol. Chem. 281(36), 26562–26568 (2006). 70. Kwon J, Lee SR, Yang KS et al. Reversible

oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors. Proc. Natl Acad. Sci. USA 101(47), 16419–16424 (2004).

future science group

PTEN & melanomagenesis

71. Leslie NR, Bennett D, Lindsay YE, Stewart

H, Gray A, Downes CP. Redox regulation of PI3-kinase signalling via inactivation of PTEN. EMBO J. 22(20), 5501–5510 (2003). 72. Lee SR, Yang KS, Kwon J, Lee C, Jeong W,

Rhee SG. Reversible inactivation of the tumor suppressor PTEN by H 2O2. J. Biol. Chem. 277(23), 20336–20342 (2002). 73. Chappell WH, Green TD, Spengeman JD,

McCubrey JA, Akula SM, Bertrand FE. Increased protein expression of the PTEN tumor suppressor in the presence of constitutively active Notch-1 Cell Cycle 4(10), 1389–1395 (2005). 74. Trotman LC, Wang X, Alimonti A et al.

Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 128(1), 141–156 (2007). 75. Wang X, Trotman LC, Koppie T et al.

NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128(1), 129–139 (2007). 76. Fouladkou F, Landry T, Kawabe H et al.

The ubiquitin ligase Nedd4-1 is dispensable for the regulation of PTEN stability and localization. Proc. Natl Acad. Sci. USA 105(25), 8585–8590 (2008). 77. Poliseno L, Salmena L, Zhang J, Carver B,

Haveman WJ, Pandolfi PP. A codingindependent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465(7301), 1033–1038 (2010). nn

Identifies a pseudogene for PTEN, which is actively transcribed, and consequently competes with the miRNA that are targeting the PTEN mRNA. It induces another level of regulation for PTEN by stabilizing the PTEN transcript.

78. Berger MF, Hodis E, Heffernan TP et al.

Melanoma genome sequencing reveals frequent PREX2 mutations. Nature 485(7399), 502–506 (2012). 79. Miller SJ, Lou DY, Seldin DC, Lane WS,

Nee LBG. Direct identification of PTEN phosphorylation sites. FEBS Lett. 528(1–3), 145–153 (2002). 80. Torres J, Pulido R. The tumor suppressor

PTEN is phosphorylated by the protein kinase CK2 at its C terminus: implications for PTEN stability to proteasome-mediated degradation. J. Biol. Chem. 276(2), 993–998 (2001). 81. Al-Khouri AM, Ma Y, Togo SH, Williams

S, Mustelin T. Cooperative phosphorylation of the tumor suppressor phosphatase and tensin homologue (PTEN) by casein kinases and glycogen synthase kinase 3beta. J. Biol. Chem. 280(42), 35195–35202 (2005).

future science group

82. Bonneau D, Longy M. Mutations of the

human PTEN gene. Hum. Mutat. 16(2), 109–122 (2000). 83. Farooq A, Walker LJ, Bowling J, Audisio

RA. Cowden syndrome. Cancer Treat. Rev. 36(8), 577–583 (2010). 84. Liaw D, Marsh DJ, Li J et al. Germline

mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat. Genet. 16(1), 64–67 (1997). 85. Yang MS, Kim CH, Cheong JH, Kim JM.

Lhermitte–Duclos disease presenting with hydrocephalus. Acta. Neurochir. Suppl. 113, 161–165 (2012). 86. Tok Celebi J, Chen FF, Zhang H, Ping XL,

Tsou HC, Peacocke M. Identification of PTEN mutations in five families with Bannayan–Zonana syndrome. Exp. Dermatol. 8(2), 134–139 (1999). 87. Virchow RLK. Cellular Pathology. John

Churchill, London, UK (1859). 88. Grille SJ, Bellacosa A, Upson J et al. The

protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63(9), 2172–2178 (2003). 89. Larue L, Bellacosa A. Epithelial–

mesenchymal transition in development and cancer. role of phosphatidylinositol 3´ kinase/AKT pathways. Oncogene 24(50), 7443–7454 (2005). 90. Kim D, Kim S, Koh H et al. Akt/PKB

promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J. 15(11), 1953–1962 (2001). 91. Henle SJ, Wang G, Liang E, Wu M, Poo

MM, Henley JR. Asymmetric PI(3,4,5)P3 and Akt signaling mediates chemotaxis of axonal growth cones. J. Neurosci. 31(19), 7016–7027 (2011). 92. Puig I, Champeval D, De Santa Barbara P,

Jaubert F, Lyonnet S, Larue L. Deletion of PTEN in the mouse enteric nervous system induces ganglioneuromatosis and mimics intestinal pseudoobstruction. J. Clin. Invest. 119(12), 3586–3596 (2009). 93. Colombo S, Champeval D, Rambow F,

Larue L. Transcriptomic analysis of mouse embryonic skin cells reveals previously unreported genes expressed in melanoblasts. J. Invest. Dermatol. 132(1), 170–178 (2012). 94. Reifenberger J, Wolter M, Boström J et al.

Allelic losses on chromosome arm 10q and mutation of the PTEN (MMAC1) tumour suppressor gene in primary and metastatic malignant melanomas. Virchows Arch. 436(5), 487–493 (2000). 95. Poetsch M, Dittberner T, Woenckhaus C.

PTEN/MMAC1 in malignant melanoma

www.futuremedicine.com

Review

and its importance for tumor progression. Cancer Genet. Cytogenet. 125(1), 21–26 (2001). 96. Guldberg P, Thor Straten P, Birck A,

Ahrenkiel V, Kirkin AF, Zeuthen J. Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event in malignant melanoma. Cancer Res. 57(17), 3660–3663 (1997). 97. Furney SJ, Turajlic S, Fenwick K et al.

Genomic characterisation of acral melanoma cell lines. Pigment Cell Melanoma Res. 25(4), 488–492 (2012). 98. Nikolaev SI, Rimoldi D, Iseli C et al. Exome

sequencing identifies recurrent somatic MAP2K1 and MAP2K2 mutations in melanoma. Nat. Genet. 44(2), 133–139 (2012). 99. You MJ, Castrillon DH, Bastian BC et al.

Genetic analysis of Pten and Ink4a/Arf interactions in the suppression of tumorigenesis in mice. Proc. Natl Acad. Sci. USA 99(3), 1455–1460 (2002). 100. Murisier F, Guichard S, Beermann F.

A conserved transcriptional enhancer that specifies Tyrp1 expression to melanocytes. Dev. Biol. 298(2), 644–655 (2006). 101. Porter S, Larue L, Mintz B. Mosaicism of

tyrosinase-locus transcription and chromatin structure in dark vs. light melanocyte clones of homozygous chinchilla-mottled mice. Dev. Genet. 12(6), 393–402 (1991). 102. Porter SD, Hu J, Gilks CB. Distal upstream

tyrosinase S/MAR-containing sequence has regulatory properties specific to subsets of melanocytes. Dev. Genet. 25(1), 40–48 (1999). 103. Delmas V, Martinozzi S, Bourgeois Y,

Holzenberger M, Larue L. Cre-mediated recombination in the skin melanocyte lineage. Genesis 36(2), 73–80 (2003). 104. Tonks ID, Nurcombe V, Paterson C et al.

Tyrosinase-Cre mice for tissue-specific gene ablation in neural crest and neuroepithelialderived tissues. Genesis 37(3), 131–138 (2003). 105. Bosenberg M, Muthusamy V, Curley DP

et al. Characterization of melanocyte-specific inducible Cre recombinase transgenic mice. Genesis 44(5), 262–267 (2006). 106. Yajima I, Belloir E, Bourgeois Y, Kumasaka

M, Delmas V, Larue L. Spatiotemporal gene control by the Cre-ERT2 system in melanocytes. Genesis 44(1), 34–43 (2006). 107. Guyonneau L, Murisier F, Rossier A, Moulin

A, Beermann F. Melanocytes and pigmentation are affected in dopachrome tautomerase knockout mice. Mol. Cell. Biol. 24(8), 3396–3403 (2004).

1119

Review

Conde-Perez & Larue

108. Inoue-Narita T, Hamada K, Sasaki T et al.

110. Damsky WE, Curley DP, Santhanakrishnan

Pten deficiency in melanocytes results in resistance to hair graying and susceptibility to carcinogen-induced melanomagenesis. Cancer Res. 68(14), 5760–5768 (2008). 109. Dankort D, Curley DP, Cartlidge RA et al.

M et al. Beta-catenin signaling controls metastasis in Braf-activated Pten-deficient melanomas. Cancer Cell 20(6), 741–754 (2011). 111. Karreth FA, Tay Y, Perna D et al. In vivo

Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 41(5), 544–552 (2009). n

Shows that BRAF and PTEN may cooperate to induce melanoma in vivo using a relevant mouse model.

1120

identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell 147(2), 382–395 (2011). n

Complementary study to [77] in which the authors demonstrate that ceRNA is regulating the PTEN mRNA.

Future Oncol. (2012) 8(9)

112. Nogueira C, Kim KH, Sung H et al.

Cooperative interactions of PTEN deficiency and RAS activation in melanoma metastasis. Oncogene 29(47), 6222–6232 (2010). 113. Longvert C, Gros G, Beermann F, Marais R,

Delmas V, Larue L. Murine cutaneous melanoma models. Importance of the genetic background. Ann. Pathol. 31(5 Suppl.), S70–S73 (2011).  ive summary

future science group